1
|
Deng RZ, Zheng X, Lu ZL, Yuan M, Meng QC, Wu T, Tian Y. Effect of colorectal cancer stem cells on the development and metastasis of colorectal cancer. World J Gastrointest Oncol 2024; 16:4354-4368. [DOI: 10.4251/wjgo.v16.i11.4354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/24/2024] [Accepted: 09/09/2024] [Indexed: 10/25/2024] Open
Abstract
The relevant mechanism of tumor-associated macrophages (TAMs) in the treatment of colorectal cancer patients with immune checkpoint inhibitors (ICIs) is discussed, and the application prospects of TAMs in reversing the treatment tolerance of ICIs are discussed to provide a reference for related studies. As a class of drugs widely used in clinical tumor immunotherapy, ICIs can act on regulatory molecules on cells that play an inhibitory role - immune checkpoints - and kill tumors in the form of an immune response by activating a variety of immune cells in the immune system. The sensitivity of patients with different types of colorectal cancer to ICI treatment varies greatly. The phenotype and function of TAMs in the colorectal cancer microenvironment are closely related to the efficacy of ICIs. ICIs can regulate the phenotypic function of TAMs, and TAMs can also affect the tolerance of colorectal cancer to ICI therapy. TAMs play an important role in ICI resistance, and making full use of this target as a therapeutic strategy is expected to improve the immunotherapy efficacy and prognosis of patients with colorectal cancer.
Collapse
Affiliation(s)
- Run-Zhi Deng
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, Fujian Province, China
| | - Xin Zheng
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, Fujian Province, China
| | - Zhong-Lei Lu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, Fujian Province, China
| | - Ming Yuan
- Department of Hepatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
| | - Qi-Chang Meng
- Department of General Surgery, Peking University First Hospital, Beijing 100034, China
| | - Tao Wu
- Department of General Surgery, West China Hospital of Sichuan University, Chengdu 610044, Sichuan Province, China
| | - Yu Tian
- Department of Thoracic Surgery, Yancheng No. 1 People’s Hospital, Affiliated Hospital of Nanjing University Medical School, The First People’s Hospital of Yancheng, Yancheng 224000, Jiangsu Province, China
| |
Collapse
|
2
|
Vitale S, Calapà F, Colonna F, Luongo F, Biffoni M, De Maria R, Fiori ME. Advancements in 3D In Vitro Models for Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405084. [PMID: 38962943 PMCID: PMC11348154 DOI: 10.1002/advs.202405084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Indexed: 07/05/2024]
Abstract
The process of drug discovery and pre-clinical testing is currently inefficient, expensive, and time-consuming. Most importantly, the success rate is unsatisfactory, as only a small percentage of tested drugs are made available to oncological patients. This is largely due to the lack of reliable models that accurately predict drug efficacy and safety. Even animal models often fail to replicate human-specific pathologies and human body's complexity. These factors, along with ethical concerns regarding animal use, urge the development of suitable human-relevant, translational in vitro models.
Collapse
Affiliation(s)
- Sara Vitale
- Department of Oncology and Molecular Medicine (OMM)Istituto Superiore di SanitàViale Regina Elena 299Rome00161Italy
| | - Federica Calapà
- Dipartimento di Medicina e Chirurgia traslazionaleUniversità Cattolica del Sacro CuoreLargo F. Vito 1RomeItaly
| | - Francesca Colonna
- Department of Oncology and Molecular Medicine (OMM)Istituto Superiore di SanitàViale Regina Elena 299Rome00161Italy
| | - Francesca Luongo
- Dipartimento di Medicina e Chirurgia traslazionaleUniversità Cattolica del Sacro CuoreLargo F. Vito 1RomeItaly
| | - Mauro Biffoni
- Department of Oncology and Molecular Medicine (OMM)Istituto Superiore di SanitàViale Regina Elena 299Rome00161Italy
| | - Ruggero De Maria
- Dipartimento di Medicina e Chirurgia traslazionaleUniversità Cattolica del Sacro CuoreLargo F. Vito 1RomeItaly
- Fondazione Policlinico Universitario “A. Gemelli” – IRCCSLargo F. Vito 1RomeItaly
| | - Micol E. Fiori
- Department of Oncology and Molecular Medicine (OMM)Istituto Superiore di SanitàViale Regina Elena 299Rome00161Italy
| |
Collapse
|
3
|
Qi F, Zhang J, Li J, Li D, Gao N, Qi Z, Kong X, Yu Z, Fang Y, Cui W, Xia J. Synergistic immunochemotherapy targeted SAMD4B-APOA2-PD-L1 axis potentiates antitumor immunity in hepatocellular carcinoma. Cell Death Dis 2024; 15:421. [PMID: 38886351 PMCID: PMC11183041 DOI: 10.1038/s41419-024-06699-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 06/20/2024]
Abstract
Targeted and immunotherapy combined with interventional therapy can improve the prognosis of advanced cancer patients, and it has become a hot spot to find the new therapeutic schemes, but most of which are not satisfactory. Single-cell RNA sequencing was performed in PDX mouse models with or without TCC therapy. 2-'O-Methylation modification and multiplex immunofluorescence staining were used to explore the function and mechanism of SAMD4B in the immune context of HCC. Here, we propose for the first time a synergistic immunochemotherapy that exerts a potent antitumour effect for patients with advanced hepatocellular carcinoma (HCC) in clinical practice based on three common antitumour drugs and found that HCC patients with new synergistic immunochemotherapy had better three-year overall survival (p = 0.004) and significantly higher survival ratio (increased by 2.3 times) than the control group. We further reveal the immunoregulatory mechanism of synergistic immunochemotherapy through 2'-O-Methylation modification mediated by SAMD4B, a tumour suppressor gene. Mechanistically, SAMD4B, increased by the reduced mutations of upstream genes NOTCH1 and NOTCH2, affected the instability of APOA2 mRNA by 2-'O-Methylation modification of the C-terminus. The decreased APOA2 further attenuated programmed death ligand 1 (PD-L1) level with a direct interaction pattern. The high-SAMD4B tumour tissues contained fewer native CD29+CD8+ T cells, which improved immune microenvironment to achieve the effect of antitumour effect. Overall, we developed a potent synergistic immunochemotherapy strategy that exerts an efficient anti-HCC effect inducing SAMD4B-APOA2-PD-L1 axis to inhibit tumour immune evasion.
Collapse
Affiliation(s)
- Feng Qi
- National Medical Center & National Clinical Research Center for Interventional Medicine. Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jian Zhang
- National Medical Center & National Clinical Research Center for Interventional Medicine. Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Jia Li
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20025, China
| | - Donghe Li
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Na Gao
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Zhuoran Qi
- National Medical Center & National Clinical Research Center for Interventional Medicine. Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Xiuyan Kong
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Zhijie Yu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Yuan Fang
- Department of Liver Surgery, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200000, China.
| | - Jinglin Xia
- National Medical Center & National Clinical Research Center for Interventional Medicine. Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
4
|
Jiang S, Zhu G, Tan Y, Zhou T, Zheng S, Wang F, Lei W, Liu X, Du J, Tian M. Identification of VEGFs-related gene signature for predicting microangiogenesis and hepatocellular carcinoma prognosis. Aging (Albany NY) 2024; 16:10321-10347. [PMID: 38874512 PMCID: PMC11236318 DOI: 10.18632/aging.205931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 04/08/2024] [Indexed: 06/15/2024]
Abstract
Microangiogenesis is an important prognostic factor in various cancers, including hepatocellular carcinoma (HCC). The Vascular Endothelial Growth Factor (VEGF) has been shown to contribute to tumor angiogenesis. Recently, several studies have investigated the regulation of VEGF production by a single gene, with few researchers exploring all genes that affect VEGF production. In this study, we comprehensively analyzed all genes affecting VEGF production in HCC and developed a risk model and gene-based risk score based on VEGF production. Moreover, the model's predictive capacity on prognosis of HCCs was verified using training and validation datasets. The developed model showed good prediction of the overall survival rate. Patients with a higher risk score experienced poor outcomes compared to those with a lower risk score. Furthermore, we identified the immunological causes of the poor prognosis of patients with high-risk scores comparing with those with low-risk scores.
Collapse
Affiliation(s)
- Shengpan Jiang
- Department of Interventional Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Guoting Zhu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Yiqing Tan
- Department of Interventional Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Tao Zhou
- Department of Interventional Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Shilin Zheng
- Department of Interventional Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Fuhua Wang
- Department of Interventional Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Wenfeng Lei
- Department of Interventional Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Xuan Liu
- Department of Interventional Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Jinjun Du
- Department of Hepatology and Gastroenterology, Wuhan Hospital of Traditional Chinese Medicine (The Third Clinical College of Hubei University of Chinese Medicine), Wuhan, Hubei Province, China
| | - Manman Tian
- Department of Hepatology and Gastroenterology, Wuhan Hospital of Traditional Chinese Medicine (The Third Clinical College of Hubei University of Chinese Medicine), Wuhan, Hubei Province, China
| |
Collapse
|
5
|
Lu W, Aihaiti A, Abudukeranmu P, Liu Y, Gao H. Unravelling the role of intratumoral bacteria in digestive system cancers: current insights and future perspectives. J Transl Med 2024; 22:545. [PMID: 38849871 PMCID: PMC11157735 DOI: 10.1186/s12967-024-05320-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 05/18/2024] [Indexed: 06/09/2024] Open
Abstract
Recently, research on the human microbiome, especially concerning the bacteria within the digestive system, has substantially advanced. This exploration has unveiled a complex interplay between microbiota and health, particularly in the context of disease. Evidence suggests that the gut microbiome plays vital roles in digestion, immunity and the synthesis of vitamins and neurotransmitters, highlighting its significance in maintaining overall health. Conversely, disruptions in these microbial communities, termed dysbiosis, have been linked to the pathogenesis of various diseases, including digestive system cancers. These bacteria can influence cancer progression through mechanisms such as DNA damage, modulation of the tumour microenvironment, and effects on the host's immune response. Changes in the composition and function within the tumours can also impact inflammation, immune response and cancer therapy effectiveness. These findings offer promising avenues for the clinical application of intratumoral bacteria for digestive system cancer treatment, including the potential use of microbial markers for early cancer detection, prognostication and the development of microbiome-targeted therapies to enhance treatment outcomes. This review aims to provide a comprehensive overview of the pivotal roles played by gut microbiome bacteria in the development of digestive system cancers. Additionally, we delve into the specific contributions of intratumoral bacteria to digestive system cancer development, elucidating potential mechanisms and clinical implications. Ultimately, this review underscores the intricate interplay between intratumoral bacteria and digestive system cancers, underscoring the pivotal role of microbiome research in transforming diagnostic, prognostic and therapeutic paradigms for digestive system cancers.
Collapse
Affiliation(s)
- Weiqin Lu
- General Surgery, Cancer Center, Department of Vascular Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | | | | | - Yajun Liu
- Aksu First People's Hospital, Xinjiang, China
| | - Huihui Gao
- Cancer Center, Department of Hospital Infection Management and Preventive Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
6
|
Dadgar N, Sherry C, Zimmerman J, Park H, Lewis C, Donnenberg A, Zaidi AH, Fan Y, Xiao K, Bartlett D, Donnenberg V, Wagner PL. Targeting interleukin-6 as a treatment approach for peritoneal carcinomatosis. J Transl Med 2024; 22:402. [PMID: 38689325 PMCID: PMC11061933 DOI: 10.1186/s12967-024-05205-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/15/2024] [Indexed: 05/02/2024] Open
Abstract
Peritoneal carcinomatosis (PC) is a complex manifestation of abdominal cancers, with a poor prognosis and limited treatment options. Recent work identifying high concentrations of the cytokine interleukin-6 (IL-6) and its soluble receptor (sIL-6-Rα) in the peritoneal cavity of patients with PC has highlighted this pathway as an emerging potential therapeutic target. This review article provides a comprehensive overview of the current understanding of the potential role of IL-6 in the development and progression of PC. We discuss mechansims by which the IL-6 pathway may contribute to peritoneal tumor dissemination, mesothelial adhesion and invasion, stromal invasion and proliferation, and immune response modulation. Finally, we review the prospects for targeting the IL-6 pathway in the treatment of PC, focusing on common sites of origin, including ovarian, gastric, pancreatic, colorectal and appendiceal cancer, and mesothelioma.
Collapse
Affiliation(s)
- Neda Dadgar
- Translational Hematology & Oncology Research, Enterprise Cancer Institute, Cleveland Clinic, Cleveland, OH, 44106, USA
| | - Christopher Sherry
- Allegheny Health Network Cancer Institute, 314 E. North Ave, Pittsburgh, PA, 15212, USA
| | - Jenna Zimmerman
- Allegheny Health Network Cancer Institute, 314 E. North Ave, Pittsburgh, PA, 15212, USA
| | - Hyun Park
- Allegheny Health Network Cancer Institute, 314 E. North Ave, Pittsburgh, PA, 15212, USA
| | - Catherine Lewis
- Allegheny Health Network Cancer Institute, 314 E. North Ave, Pittsburgh, PA, 15212, USA
| | - Albert Donnenberg
- Allegheny Health Network Cancer Institute, 314 E. North Ave, Pittsburgh, PA, 15212, USA
| | - Ali H Zaidi
- Allegheny Health Network Cancer Institute, 314 E. North Ave, Pittsburgh, PA, 15212, USA
| | - Yong Fan
- Allegheny Health Network Cancer Institute, 314 E. North Ave, Pittsburgh, PA, 15212, USA
| | - Kunhong Xiao
- Center for Proteomics & Artificial Intelligence, Center for Clinical Mass Spectrometry, Allegheny Health Network Cancer Institute, Pittsburgh, PA, 15224, USA
| | - David Bartlett
- Allegheny Health Network Cancer Institute, 314 E. North Ave, Pittsburgh, PA, 15212, USA
| | - Vera Donnenberg
- University of Pittsburgh School of MedicineDepartment of Cardiothoracic SurgeryUPMC Hillman Cancer Center Wagner, Patrick; Allegheny Health Network Cancer Institute, Pittsburgh, USA
| | - Patrick L Wagner
- Allegheny Health Network Cancer Institute, 314 E. North Ave, Pittsburgh, PA, 15212, USA.
| |
Collapse
|
7
|
Yan J, Zhu J, Li X, Yang R, Xiao W, Huang C, Zheng C. Blocking LTB 4 signaling-mediated TAMs recruitment by Rhizoma Coptidis sensitizes lung cancer to immunotherapy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 119:154968. [PMID: 37531900 DOI: 10.1016/j.phymed.2023.154968] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 05/31/2023] [Accepted: 07/15/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND Immune checkpoint blockade (ICB) induces durable immune responses across a spectrum of advanced cancers and revolutionizes the oncology field. However, only a subset of patients achieves long-lasting clinical benefits. Tumor-associated macrophages (TAMs) usually secrete immunosuppressive cytokines and contribute to the failure of ICB therapy. Therefore, it is crucial to mechanically manipulate the abundance and function of TAMs in the tumor microenvironment (TME), which can offer a promising molecular basis to improve the clinical response efficacy of ICB in cancer patients. PURPOSE This study aims to investigate TAMs in the immunosuppressive microenvironment to identify new therapeutic targets, improve the ability to predict and guide responses to clinical immunotherapy, and develop new strategies for immunotherapy of lung tumors. METHODS Lewis lung carcinoma (LLC) xenograft-bearing mouse models were established to analyze the antitumor activity of Rhizoma Coptidis (RC) in vivo. A systems pharmacology strategy was used to predict the correlation between RC and M2 macrophages. The effect of RC on the abundance of M2 macrophages was analyzed by flow cytometry of murine samples. Western blot was performed to analyze the expression of Leukotriene A4 hydrolase (LTA4H) and LTB4 receptor 1 (BLT1) in harvested lung cancer tissues. The impact of blocking leukotriene B4 (LTB4) signaling by RC on the recruitment of M2 macrophages was assessed in vitro and in vivo. Transwell migration assays were conducted to clarify the inhibition of macrophage migration by blocking LTB4. Lta4h-/- mice were used to investigate the sensitivity of immunotherapy to lung cancer by blocking the LTB4 signaling. RESULTS Here, we report that RC, an herbal medicine from the family Ranunculaceae, suppresses the recruitment and immunosuppressive function of TAMs, which in turn sensitizes lung cancer to ICB therapy. Firstly, a systems pharmacology strategy was proposed to identify combinatorial drugs for ICB therapy with a systems biology perspective of drug-target-pathway-TME phenotype. We predicted and verified that RC significantly inhibits tumor growth and the infiltration of M2-TAMs into TME of LLC tumor-bearing mice. Then, RC inhibits the recruitment of macrophages to the tumor TME via blocking LTB4 signaling, and suppresses the expression of immunosuppressive factors (IL-10, TGF-β and VEGF). As a result, RC enables CD8+ T cells to retain their proliferative and infiltrative abilities within the TME. Ultimately, these events promote cytotoxic T-cell-mediated clearance of tumor cells, which is further enhanced by the addition of anti-PD-L1 therapy. Furthermore, we employed LTA4H deficient mice (Lta4h-/- mice) to evaluate the antitumor efficiency, the results showed that the efficacy of immunotherapy was enhanced due to the synergistic effect of LTB4 signaling blockage and ICB inhibition, leading to remarkable inhibition of tumor growth in a mouse model of lung adenocarcinoma. CONCLUSIONS Taken together, these findings suggest that RC enhances antitumor immunity, providing a rationale for combining RC with immunotherapies as a potential anti-cancer treatment strategy.
Collapse
Affiliation(s)
- Jiangna Yan
- College of Medicine, Yan'an University, Yan'an, Shaanxi 716000, PR China
| | - Jinglin Zhu
- College of Medicine, Yan'an University, Yan'an, Shaanxi 716000, PR China
| | - Xiaolan Li
- Department of Pathology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi 710038, PR China
| | - Ruijie Yang
- School of Life Sciences, East China Normal University, Shanghai 200241, PR China
| | - Wei Xiao
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Parmaceutical Co. Ltd., Lianyungang, Jiangsu 222001, PR China
| | - Chao Huang
- School of Basic Medical Sciences, Institute of Molecular and Translational Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China.
| | - Chunli Zheng
- College of Medicine, Yan'an University, Yan'an, Shaanxi 716000, PR China.
| |
Collapse
|
8
|
Gu J, Lv X, Li W, Li G, He X, Zhang Y, Shi L, Zhang X. Deciphering the mechanism of Peptostreptococcus anaerobius-induced chemoresistance in colorectal cancer: the important roles of MDSC recruitment and EMT activation. Front Immunol 2023; 14:1230681. [PMID: 37781363 PMCID: PMC10533913 DOI: 10.3389/fimmu.2023.1230681] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/24/2023] [Indexed: 10/03/2023] Open
Abstract
Peptostreptococcus anaerobius (P. anaerobius, PA) in intestinal flora of patients with colorectal cancer (CRC) are associated with poor prognosis. Studies have shown that P. anaerobius could promote colorectal carcinogenesis and progression, but whether P. anaerobius could induce chemoresistance of colorectal cancer has not been clarified. Here, both in vitro and in vivo experiments showed that P. anaerobius specifically colonized the CRC lesion and enhanced chemoresistance of colorectal cancer to oxaliplatin by recruiting myeloid-derived suppressor cells (MDSCs) into the tumor microenvironment. Furthermore, this study revealed that it was the increased secretion of IL-23 by MDSCs that subsequently facilitated the epithelial-mesenchymal transition (EMT) of tumor cells to induce chemoresistance of CRC by activating the Stat3-EMT pathway. Our results highlight that targeting P. anaerobius might be a novel therapeutic strategy to overcome chemoresistance in the treatment of CRC.
Collapse
Affiliation(s)
- Jinhua Gu
- Department of Gastroenterology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xiaojun Lv
- Department of Gastroenterology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Wenwen Li
- Department of Gastroenterology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Guangcai Li
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Xialian He
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Ye Zhang
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Lihong Shi
- College of Rehabilitation Medicine, Weifang Medical University, Weifang, China
| | - Xiaoqian Zhang
- Department of Gastroenterology, Affiliated Hospital of Weifang Medical University, Weifang, China
| |
Collapse
|
9
|
Chen L, Gao W, Lin L, Sha C, Li T, Chen Q, Wei H, Yang M, Xing J, Zhang M, Zhao S, Xu W, Li Y, Long L, Zhu X. A methylation- and immune-related lncRNA signature to predict ovarian cancer outcome and uncover mechanisms of chemoresistance. J Ovarian Res 2023; 16:186. [PMID: 37674251 PMCID: PMC10483746 DOI: 10.1186/s13048-023-01260-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 08/13/2023] [Indexed: 09/08/2023] Open
Abstract
Tumor-associated lncRNAs regulated by epigenetic modification switches mediate immune escape and chemoresistance in ovarian cancer (OC). However, the underlying mechanisms and concrete targets have not been systematically elucidated. Here, we discovered that methylation modifications played a significant role in regulating immune cell infiltration and sensitizing OC to chemotherapy by modulating immune-related lncRNAs (irlncRNAs), which represent tumor immune status. Through deep analysis of the TCGA database, a prognostic risk model incorporating four methylation-related lncRNAs (mrlncRNAs) and irlncRNAs was constructed. Twenty-one mrlncRNA/irlncRNA pairs were identified that were significantly related to the overall survival (OS) of OC patients. Subsequently, we selected four lncRNAs to construct a risk signature predictive of OS and indicative of OC immune infiltration, and verified the robustness of the risk signature in an internal validation set. The risk score was an independent prognostic factor for OC prognosis, which was demonstrated via multifactorial Cox regression analysis and nomogram. Moreover, risk scores were negatively related to the expression of CD274, CTLA4, ICOS, LAG3, PDCD1, and PDCD1LG2 and negatively correlated with CD8+, CD4+, and Treg tumor-infiltrating immune cells. In addition, a high-risk score was associated with a higher IC50 value for cisplatin, which was associated with a significantly worse clinical outcome. Next, a competing endogenous RNA (ceRNA) network and a signaling pathway controlling the infiltration of CD8+ T cells were explored based on the lncRNA model, which suggested a potential therapeutic target for immunotherapy. Overall, this study constructed a prognostic model by pairing mrlncRNAs and irlncRNAs and revealed the critical role of the FTO/RP5-991G20.1/hsa-miR-1976/MEIS1 signaling pathway in regulating immune function and enhancing anticancer therapy.
Collapse
Affiliation(s)
- Lu Chen
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
- Department of Gynaecology and Obstetrics, Taixing People's Hospital, Taixing, Jiangsu, China
| | - Wujiang Gao
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
- Department of Gynaecology and Obstetrics, Yangzhou First People's Hospital, Yangzhou, Jiangsu, China
| | - Li Lin
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
| | - Chunli Sha
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
- Department of Gynaecology and Obstetrics, The First People's Hospital of Nantong City, Nantong, Jiangsu, China
| | - Taoqiong Li
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
| | - Qi Chen
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
| | - Hong Wei
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
| | - Meiling Yang
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
- Department of Gynaecology and Obstetrics, The First People's Hospital of Nantong City, Nantong, Jiangsu, China
| | - Jie Xing
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
| | - Mengxue Zhang
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
| | - Shijie Zhao
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
| | - Wenlin Xu
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China.
| | - Yuefeng Li
- Medical school, Jiangsu University, No. 301, Xuefu Road, Zhenjiang City, 212031, Jiangsu Province, China.
| | - Lulu Long
- Oncology Department, Affiliated People's Hospital of jiangsu university, No. 8, Dianli Road, Zhenjiang City, 212001, Jiangsu Province, China.
| | - Xiaolan Zhu
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China.
| |
Collapse
|
10
|
Geng T, Zheng M, Wang Y, Reseland JE, Samara A. An artificial intelligence prediction model based on extracellular matrix proteins for the prognostic prediction and immunotherapeutic evaluation of ovarian serous adenocarcinoma. Front Mol Biosci 2023; 10:1200354. [PMID: 37388244 PMCID: PMC10301747 DOI: 10.3389/fmolb.2023.1200354] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/31/2023] [Indexed: 07/01/2023] Open
Abstract
Background: Ovarian Serous Adenocarcinoma is a malignant tumor originating from epithelial cells and one of the most common causes of death from gynecological cancers. The objective of this study was to develop a prediction model based on extracellular matrix proteins, using artificial intelligence techniques. The model aimed to aid healthcare professionals to predict the overall survival of patients with ovarian cancer (OC) and determine the efficacy of immunotherapy. Methods: The Cancer Genome Atlas Ovarian Cancer (TCGA-OV) data collection was used as the study dataset, whereas the TCGA-Pancancer dataset was used for validation. The prognostic importance of 1068 known extracellular matrix proteins for OC were determined by the Random Forest algorithm and the Lasso algorithm establishing the ECM risk score. Based on the gene expression data, the differences in mRNA abundance, tumour mutation burden (TMB) and tumour microenvironment (TME) between the high- and low-risk groups were assessed. Results: Combining multiple artificial intelligence algorithms we were able to identify 15 key extracellular matrix genes, namely, AMBN, CXCL11, PI3, CSPG5, TGFBI, TLL1, HMCN2, ESM1, IL12A, MMP17, CLEC5A, FREM2, ANGPTL4, PRSS1, FGF23, and confirm the validity of this ECM risk score for overall survival prediction. Several other parameters were identified as independent prognostic factors for OC by multivariate COX analysis. The analysis showed that thyroglobulin (TG) targeted immunotherapy was more effective in the high ECM risk score group, while the low ECM risk score group was more sensitive to the RYR2 gene-related immunotherapy. Additionally, the patients with low ECM risk scores had higher immune checkpoint gene expression and immunophenoscore levels and responded better to immunotherapy. Conclusion: The ECM risk score is an accurate tool to assess the patient's sensitivity to immunotherapy and forecast OC prognosis.
Collapse
Affiliation(s)
- Tianxiang Geng
- Department of Biomaterials, FUTURE, Center for Functional Tissue Reconstruction, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Mengxue Zheng
- Laboratory of Reproductive Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yongfeng Wang
- Department of Obstetrics and Gynecology, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Janne Elin Reseland
- Department of Biomaterials, FUTURE, Center for Functional Tissue Reconstruction, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Athina Samara
- Department of Biomaterials, FUTURE, Center for Functional Tissue Reconstruction, Faculty of Dentistry, University of Oslo, Oslo, Norway
| |
Collapse
|
11
|
Wu T, Zhang X, Liu X, Cai X, Shen T, Pan D, Liang R, Ding R, Hu R, Dong J, Li F, Li J, Xie L, Wang C, Geng S, Yang Z, Xing L, Li Y. Single-cell sequencing reveals the immune microenvironment landscape related to anti-PD-1 resistance in metastatic colorectal cancer with high microsatellite instability. BMC Med 2023; 21:161. [PMID: 37106440 PMCID: PMC10142806 DOI: 10.1186/s12916-023-02866-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND The objective response rate of microsatellite instability-high (MSI-H) metastatic colorectal cancer (mCRC) patients with first-line anti-programmed cell death protein-1 (PD-1) monotherapy is only 40-45%. Single-cell RNA sequencing (scRNA-seq) enables unbiased analysis of the full variety of cells comprising the tumor microenvironment. Thus, we used scRNA-seq to assess differences among microenvironment components between therapy-resistant and therapy-sensitive groups in MSI-H/mismatch repair-deficient (dMMR) mCRC. Resistance-related cell types and genes identified by this analysis were subsequently verified in clinical samples and mouse models to further reveal the molecular mechanism of anti-PD-1 resistance in MSI-H or dMMR mCRC. METHODS The response of primary and metastatic lesions to first-line anti-PD-1 monotherapy was evaluated by radiology. Cells from primary lesions of patients with MSI-H/dMMR mCRC were analyzed using scRNA-seq. To identify the marker genes in each cluster, distinct cell clusters were identified and subjected to subcluster analysis. Then, a protein‒protein interaction network was constructed to identify key genes. Immunohistochemistry and immunofluorescence were applied to verify key genes and cell marker molecules in clinical samples. Immunohistochemistry, quantitative real-time PCR, and western blotting were performed to examine the expression of IL-1β and MMP9. Moreover, quantitative analysis and sorting of myeloid-derived suppressor cells (MDSCs) and CD8+ T cells were performed using flow cytometry. RESULTS Tumor responses in 23 patients with MSI-H/dMMR mCRC were evaluated by radiology. The objective response rate was 43.48%, and the disease control rate was 69.57%. ScRNA-seq analysis showed that, compared with the treatment-resistant group, the treatment-sensitive group accumulated more CD8+ T cells. Experiments with both clinical samples and mice indicated that infiltration of IL-1β-driven MDSCs and inactivation of CD8+ T cells contribute to anti-PD-1 resistance in MSI-H/dMMR CRC. CONCLUSIONS CD8+ T cells and IL-1β were identified as the cell type and gene, respectively, with the highest correlation with anti-PD-1 resistance. Infiltration of IL-1β-driven MDSCs was a significant factor in anti-PD-1 resistance in CRC. IL-1β antagonists are expected to be developed as a new treatment for anti-PD-1 inhibitor resistance.
Collapse
Affiliation(s)
- Tao Wu
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, No. 519, Kunzhou Road, Xishan District, Kunming, 650118, China
| | - Xuan Zhang
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, No. 519, Kunzhou Road, Xishan District, Kunming, 650118, China
| | - Xinxing Liu
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, No. 519, Kunzhou Road, Xishan District, Kunming, 650118, China
| | - Xinyi Cai
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, No. 519, Kunzhou Road, Xishan District, Kunming, 650118, China
| | - Tao Shen
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, No. 519, Kunzhou Road, Xishan District, Kunming, 650118, China
| | - Dingguo Pan
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, No. 519, Kunzhou Road, Xishan District, Kunming, 650118, China
| | - Rui Liang
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Rong Ding
- Department of Minimally Invasive Intervention, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ruixi Hu
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, No. 519, Kunzhou Road, Xishan District, Kunming, 650118, China
| | - Jianhua Dong
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, No. 519, Kunzhou Road, Xishan District, Kunming, 650118, China
| | - Furong Li
- Department of Gastroenteroscopy, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jinsha Li
- Department of Gastroenteroscopy, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lin Xie
- Department of Oncology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Chunlong Wang
- Department of Radiology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shilei Geng
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, No. 519, Kunzhou Road, Xishan District, Kunming, 650118, China
| | - Zhaoyu Yang
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, No. 519, Kunzhou Road, Xishan District, Kunming, 650118, China
| | - Lu Xing
- Department of Dermatology, Kunming Children's Hospital, Kunming, China.
| | - YunFeng Li
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, No. 519, Kunzhou Road, Xishan District, Kunming, 650118, China.
| |
Collapse
|
12
|
Li X, Li L, Xiong X, Kuang Q, Peng M, Zhu K, Luo P. Identification of the Prognostic Biomarkers CBX6 and CBX7 in Bladder Cancer. Diagnostics (Basel) 2023; 13:diagnostics13081393. [PMID: 37189494 DOI: 10.3390/diagnostics13081393] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/22/2023] [Accepted: 04/07/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Chromobox (CBX) proteins are essential components of polycomb group proteins and perform essential functions in bladder cancer (BLCA). However, research on CBX proteins is still limited, and the function of CBXs in BLCA has not been well illustrated. METHODS AND RESULTS We analyzed the expression of CBX family members in BLCA patients from The Cancer Genome Atlas database. By Cox regression analysis and survival analysis, CBX6 and CBX7 were identified as potential prognostic factors. Subsequently, we identified genes associated with CBX6/7 and performed enrichment analysis, and they were enriched in urothelial carcinoma and transitional carcinoma. Mutation rates of TP53 and TTN correlate with expression of CBX6/7. In addition, differential analysis indicated that the roles played by CBX6 and CBX7 may be related to immune checkpoints. The CIBERSORT algorithm was used to screen out immune cells that play a role in the prognosis of bladder cancer patients. Multiplex immunohistochemistry staining confirmed a negative correlation between CBX6 and M1 macrophages, as well as a consistent alteration in CBX6 and regulatory T cells (Tregs), a positive correlation between CBX7 and resting mast cells, and a negative correlation between CBX7 and M0 macrophages. CONCLUSIONS CBX6 and CBX7 expression levels may assist in predicting the prognosis of BLCA patients. CBX6 may contribute to a poor prognosis in patients by inhibiting M1 polarization and promoting Treg recruitment in the tumor microenvironment, while CBX7 may contribute to a better prognosis in patients by increasing resting mast cell numbers and decreasing macrophage M0 content.
Collapse
Affiliation(s)
- Xinxin Li
- Department of Urology, Wuhan Third Hospital and Tongren Hospital of Wuhan University, Wuhan 430060, China
| | - Lili Li
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xi Xiong
- Department of Urology, Wuhan Third Hospital, School of Medicine, Wuhan University of Science and Technology, Wuhan 430060, China
| | - Qihui Kuang
- Department of Urology, Wuhan Third Hospital and Tongren Hospital of Wuhan University, Wuhan 430060, China
| | - Min Peng
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Kai Zhu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Pengcheng Luo
- Department of Urology, Wuhan Third Hospital and Tongren Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
13
|
Chen R, Wei JM. Integrated analysis identifies oxidative stress-related lncRNAs associated with progression and prognosis in colorectal cancer. BMC Bioinformatics 2023; 24:76. [PMID: 36869292 PMCID: PMC9985255 DOI: 10.1186/s12859-023-05203-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 02/23/2023] [Indexed: 03/05/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common cancers in the world. Oxidative stress reactions have been reportedly associated with oncogenesis and tumor progression. By analyzing mRNA expression data and clinical information from The Cancer Genome Atlas (TCGA), we aimed to construct an oxidative stress-related long noncoding RNA (lncRNA) risk model and identify oxidative stress-related biomarkers to improve the prognosis and treatment of CRC. RESULTS Differentially expressed oxidative stress-related genes (DEOSGs) and oxidative stress-related lncRNAs were identified by using bioinformatics tools. An oxidative stress-related lncRNA risk model was constructed based on 9 lncRNAs (AC034213.1, AC008124.1, LINC01836, USP30-AS1, AP003555.1, AC083906.3, AC008494.3, AC009549.1, and AP006621.3) by least absolute shrinkage and selection operator (LASSO) analysis. The patients were then divided into high- and low-risk groups based on the median risk score. The high-risk group had a significantly worse overall survival (OS) (p < 0.001). Receiver operating characteristic (ROC) and calibration curves displayed the favorable predictive performance of the risk model. The nomogram successfully quantified the contribution of each metric to survival, and the concordance index and calibration plots demonstrated its excellent predictive capacity. Notably, different risk subgroups showed significant differences in terms of their metabolic activity, mutation landscape, immune microenvironment and drug sensitivity. Specifically, differences in the immune microenvironment implied that CRC patients in certain subgroups might be more responsive to immune checkpoint inhibitors. CONCLUSIONS Oxidative stress-related lncRNAs can predict the prognosis of CRC patients, which provides new insight for future immunotherapies based on potential oxidative stress targets.
Collapse
Affiliation(s)
- Rui Chen
- Department of Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jun-Min Wei
- Department of Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
14
|
Zeng C, He R, Dai Y, Lu X, Deng L, Zhu Q, Liu Y, Liu Q, Lu W, Wang Y, Jin J. Identification of TGF-β signaling-related molecular patterns, construction of a prognostic model, and prediction of immunotherapy response in gastric cancer. Front Pharmacol 2022; 13:1069204. [PMID: 36467074 PMCID: PMC9715605 DOI: 10.3389/fphar.2022.1069204] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/07/2022] [Indexed: 06/22/2024] Open
Abstract
Background: TGF-β signaling pathway plays an essential role in tumor progression and immune responses. However, the link between TGF-β signaling pathway-related genes (TSRGs) and clinical prognosis, tumor microenvironment (TME), and immunotherapy in gastric cancer is unclear. Methods: Transcriptome data and related clinical data of gastric cancer were downloaded from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, and 54 TSRGs were obtained from the Molecular Signatures Database (MSigDB). We systematically analyzed the expression profile characteristics of 54 TSRGs in 804 gastric cancer samples and examined the differences in prognosis, clinicopathological features, and TME among different molecular subtypes. Subsequently, TGF-β-related prognostic models were constructed using univariate and least absolute shrinkage and selection operator (LASSO) Cox regression analysis to quantify the degree of risk in each patient. Patients were divided into two high- and low-risk groups based on the median risk score. Finally, sensitivity to immune checkpoint inhibitors (ICIs) and anti-tumor agents was assessed in patients in high- and low-risk groups. Results: We identified two distinct TGF-β subgroups. Compared to TGF-β cluster B, TGF-β cluster A exhibits an immunosuppressive microenvironment with a shorter overall survival (OS). Then, a novel TGF-β-associated prognostic model, including SRPX2, SGCE, DES, MMP7, and KRT17, was constructed, and the risk score was demonstrated as an independent prognostic factor for gastric cancer patients. Further studies showed that gastric cancer patients in the low-risk group, characterized by higher tumor mutation burden (TMB), the proportion of high microsatellite instability (MSI-H), immunophenoscore (IPS), and lower tumor immune dysfunction and exclusion (TIDE) score, had a better prognosis, and linked to higher response rate to immunotherapy. In addition, the risk score and anti-tumor drug sensitivity were strongly correlated. Conclusion: These findings highlight the importance of TSRGs, deepen the understanding of tumor immune microenvironment, and guide individualized immunotherapy for gastric cancer patients.
Collapse
Affiliation(s)
- Cheng Zeng
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
- Department of Oncology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | - Rong He
- Department of Medical Oncology, Shanghai Tenths People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuyang Dai
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xiaohuan Lu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Linghui Deng
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
- Department of Oncology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qi Zhu
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
- Department of Oncology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | - Yu Liu
- Department of Internal Medicine, School of Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Qian Liu
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
- Department of Oncology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | - Wenbin Lu
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
- Department of Oncology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | - Yue Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jianhua Jin
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
- Department of Oncology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| |
Collapse
|
15
|
Ding YN, Xue M, Tang QS, Wang LJ, Ding HY, Li H, Gao CC, Yu WP. Immunotherapy-based novel nanoparticles in the treatment of gastrointestinal cancer: Trends and challenges. World J Gastroenterol 2022; 28:5403-5419. [PMID: 36312831 PMCID: PMC9611702 DOI: 10.3748/wjg.v28.i37.5403] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/27/2022] [Accepted: 09/15/2022] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal cancer (GIC) is the most common cancer with a poor prognosis. Currently, surgery is the main treatment for GIC. However, the high rate of postoperative recurrence leads to a low five-year survival rate. In recent years, immunotherapy has received much attention. As the only immunotherapy drugs approved by the Food and Drug Administration (FDA), immune checkpoint blockade (ICB) drugs have great potential in cancer therapy. Nevertheless, the efficacy of ICB treatment is greatly limited by the low immunogenicity and immunosuppressive microenvironment of GIC. Therefore, the targets of immunotherapy have expanded from ICB to increasing tumor immunogenicity, increasing the recruitment and maturation of immune cells and reducing the proportion of inhibitory immune cells, such as M2-like macrophages, regulatory T cells and myeloid-derived suppressor cells. Moreover, with the development of nanotechnology, a variety of nanoparticles have been approved by the FDA for clinical therapy, so novel nanodrug delivery systems have become a research focus for anticancer therapy. In this review, we summarize recent advances in the application of immunotherapy-based nanoparticles in GICs, such as gastric cancer, hepatocellular carcinoma, colorectal cancer and pancreatic cancer, and described the existing challenges and future trends.
Collapse
Affiliation(s)
- Yi-Nan Ding
- Department of Pathophysiology, College of Medicine, Southeast University, Nanjing 210000, Jiangsu Province, China
| | - Ming Xue
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210000, Jiangsu Province, China
| | - Qiu-Sha Tang
- Department of Pathophysiology, College of Medicine, Southeast University, Nanjing 210000, Jiangsu Province, China
| | - Li-Jun Wang
- Department of Pathophysiology, College of Medicine, Southeast University, Nanjing 210000, Jiangsu Province, China
| | - Hui-Yan Ding
- Department of Pathophysiology, College of Medicine, Southeast University, Nanjing 210000, Jiangsu Province, China
| | - Han Li
- Department of Tuberculosis, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Cheng-Cheng Gao
- Department of Radiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Wei-Ping Yu
- Medical School, Southeast University, Nanjing 210009, Jiangsu Province, China
| |
Collapse
|
16
|
Wang H, Pan W. Challenges of chimeric antigen receptor-T/natural killer cell therapy in the treatment of solid tumors: focus on colorectal cancer and evaluation of combination therapies. Mol Cell Biochem 2022; 478:967-980. [PMID: 36190614 DOI: 10.1007/s11010-022-04568-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 09/16/2022] [Indexed: 11/25/2022]
Abstract
Colorectal cancer (CRC) is the second most common cancer globally and one of the deadliest human malignancies. Traditional therapies, such as surgery, chemotherapy, and combination therapies have been used to treat patients with CRC. However, recently immunotherapy has been considered a practical and attractive therapeutic approach in various cancers, such as CRC. Among the immunotherapy methods, chimeric antigen receptor (CAR)-T, and CAR-natural killer cells (NK) cells therapy have been significantly successful, mainly in treating hematological malignancies. However, the effectiveness of CAR-T/NK cell therapy in the treatment of solid tumors, such as CRC has been less than blood malignancies due to various challenges, such as the selection of tumor antigens, lack of proper trafficking in tumor tissue, immunosuppressive tumor microenvironment, tumor heterogeneity and, adverse effects during and after CAR-T/NK cell therapy. This review summarized the biological structure of CAR-T/NK cells and their use in various types of human malignancies, particularly CRC, as well as the challenges of this type of treatment and the outcome of related combination therapies.
Collapse
Affiliation(s)
- Haifeng Wang
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, 312000, China
| | - Weihuo Pan
- Department of Colorectal Surgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, 568# Zhongxing North Road, Shaoxing, 312000, China.
| |
Collapse
|
17
|
OuYang LY, Deng ZJ, You YF, Fang JM, Chen XJ, Liu JJ, Li XZ, Lian L, Chen S. SIRGs score may be a predictor of prognosis and immunotherapy response for esophagogastric junction adenocarcinoma. Front Immunol 2022; 13:977894. [PMID: 36052090 PMCID: PMC9424497 DOI: 10.3389/fimmu.2022.977894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 07/22/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundEsophagogastric junction adenocarcinoma (EGJA) is a special malignant tumor with unknown biological behavior. PD-1 checkpoint inhibitors have been recommended as first-line treatment for advanced EGJA patients. However, the biomarkers for predicting immunotherapy response remain controversial.MethodsWe identified stromal immune-related genes (SIRGs) by ESTIMATE from the TCGA-EGJA dataset and constructed a signature score. In addition, survival analysis was performed in both the TCGA cohort and GEO cohort. Subsequently, we explored the differences in tumor-infiltrating immune cells, immune subtypes, immune-related functions, tumor mutation burden (TMB), immune checkpoint gene expression, immunophenoscore (IPS) between the high SIRGs score and low SIRGs score groups. Finally, two validation cohorts of patients who had accepted immunotherapy was used to verify the value of SIRGs score in predicting immunotherapy response.ResultsEight of the SIRGs were selected by LASSO regression to construct a signature score (SIRGs score). Univariate and multivariate analyses in the TCGA and GEO cohort suggested that SIRGs score was an independent risk factor for the overall survival (OS) and it could increase the accuracy of clinical prediction models for survival. However, in the high SIRGs score group, patients had more immune cell infiltration, more active immune-related functions, higher immune checkpoint gene expression and higher IPS-PD1 and IPS-PD1-CTLA4 scores, which indicate a better response to immunotherapy. The external validation illustrated that high SIRGs score was significantly associated with immunotherapy response and immune checkpoint inhibitors (ICIs) can improve OS in patients with high SIRGs score.ConclusionThe SIRGs score may be a predictor of the prognosis and immune-therapy response for esophagogastric junction adenocarcinoma.
Collapse
Affiliation(s)
- Li-Ying OuYang
- Department of Intensive Care Unit, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zi-Jian Deng
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
| | - Yu-Feng You
- School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jia-Ming Fang
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
| | - Xi-Jie Chen
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
| | - Jun-Jie Liu
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
| | - Xian-Zhe Li
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
| | - Lei Lian
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
| | - Shi Chen
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
- *Correspondence: Shi Chen,
| |
Collapse
|
18
|
The Role of Inflammatory Cytokines in the Pathogenesis of Colorectal Carcinoma—Recent Findings and Review. Biomedicines 2022; 10:biomedicines10071670. [PMID: 35884974 PMCID: PMC9312930 DOI: 10.3390/biomedicines10071670] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 11/17/2022] Open
Abstract
The inflammatory process plays a significant role in the development of colon cancer (CRC). Intestinal cytokine networks are critical mediators of tissue homeostasis and inflammation but also impact carcinogenesis at all stages of the disease. Recent studies suggest that inflammation is of greater importance in the serrated pathway than in the adenoma-carcinoma pathway. Interleukins have gained the most attention due to their potential role in CRC pathogenesis and promising results of clinical trials. Malignant transformation is associated with the pro-tumorigenic and anti-tumorigenic cytokines. The harmony between proinflammatory and anti-inflammatory factors is crucial to maintaining homeostasis. Immune cells in the tumor microenvironment modulate immune sensitivity and facilitate cancer escape from immune surveillance. Therefore, clarifying the role of underlying cytokine pathways and the effects of their modulation may be an important step to improve the effectiveness of cancer immunotherapy.
Collapse
|
19
|
Liu X, Song Q, Wang D, Liu Y, Zhang Z, Fu W. LIMK1: A promising prognostic and immune infiltration indicator in colorectal cancer. Oncol Lett 2022; 24:234. [PMID: 35720504 PMCID: PMC9185146 DOI: 10.3892/ol.2022.13354] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/10/2022] [Indexed: 12/09/2022] Open
Abstract
Studies have shown that LIM domain kinase 1 (LIMK1) is upregulated in a variety of tumors and may be a potential detection target. The present study analyzed the expression difference of LIMK1 and its relationship with tumor clinicopathological characteristics and tumor microenvironment in colorectal cancer (CRC). The transcriptomic data of LIMK1 with CRC were downloaded from The Cancer Genome Atlas (TCGA) database and GEO databases for analyzing the expression of LIMK1 mRNA and the correlation with the prognosis of patients. The protein expression of LIMK1 was obtained from the Human Protein Atlas. The receiver operating characteristic (ROC) curve and Kaplan-Meier was used to evaluate the expression characteristics and prognostic differences of LIMK1 in CRC. STRING was used to analyze co-expression genes of LIMK1. The tumor immune estimation resource was applied to the correlation between LIMK1 expression and immune infiltrates. The present study verified LIMK1 expression at the level of clinical samples collected from the Tianjin Medical University General Hospital and cell lines using reverse transcription-quantitative PCR. The mRNA and protein expression of LIMK1 were both upregulated in tumor tissues compared with adjacent tissues in CRC. The expression levels of LIMK1 were positively associated with clinical-pathological features of CRC including lymphatic invasion (P=4.00×10−2) and high pathologic stages (P=4.20×10−2). The AUC value of LIMK1 in CRC was 0.937 (95% CI: 0.918-0.957) through ROC analysis. Under the best cut-off value (4.009), the sensitivity and specificity were 98 and 81.9%. LIMK1 expression was mainly related to CD4+ T cells, macrophages and dendritic cells in the immune microenvironment of CRC. In conclusion, the high expression of LIMK1 in CRC was closely related to the clinical features and prognosis of patients. Therefore, LIMK1 was a promising prognostic indicator and a potential target for immunotherapy in CRC.
Collapse
Affiliation(s)
- Xin Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Qiang Song
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Daohan Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yubiao Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Zhixiang Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Weihua Fu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
20
|
Guo Y, Li G, Xu M, Peng B. A lncRNA signature of tumor-infiltrating macrophages is associated with prognosis and tumor immunity in lung adenocarcinoma. Comput Biol Med 2022; 148:105655. [PMID: 35715263 DOI: 10.1016/j.compbiomed.2022.105655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/05/2022] [Accepted: 05/18/2022] [Indexed: 12/09/2022]
Abstract
Accumulating evidence suggests that long noncoding RNAs (lncRNAs) play a critical role in regulating cancer immunity and the tumor microenvironment. Tumor-infiltrating macrophages are one of the most abundant constituents of many tumors. However, the functions and clinical significance of lncRNAs in tumor-associated macrophages have not been systematically elucidated. In this study, we analyzed the tumor immune microenvironment and lncRNA expression level differences based on The Cancer Genome Atlas (TCGA) and immune cell transcriptome profiles using lung adenocarcinoma microarray datasets GSE3141, GSE51210, GSE37745, and the RNA sequencing data of GSE81089. We then identified a macrophage infiltration-related lncRNA signature (MILnc) including LINC00240, MCF2L-AS1, SFTA3, MIR497HG, FAM215A, UCA1, MIR155HG, and TLRB-AS1 from a list of 147 macrophage-specific lncRNAs. The MILnc was capable of predicting overall survival differences in TCGA and several external validation datasets with a favorable performance. Functional analysis revealed that MILnc was associated with tumor progression and negatively correlated with immune checkpoints. Additionally, MILnc was positively correlated with tumor mutational burden and could predict the immunotherapy response of patients receiving anti-PD-1 or anti-CTLA4 therapy. In summary, our study highlighted the value of MILnc, which revealed the immune environment status and immunotherapy response of lung adenocarcinoma. A robust and powerful MILnc risk model could aid exploration of treatment decisions and mechanisms of macrophage-infiltrating lncRNAs.
Collapse
Affiliation(s)
- Yanhua Guo
- Department of Thoracic Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), 518020, Shenzhen, China
| | - Guofeng Li
- Department of Thoracic Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), 518020, Shenzhen, China
| | - Mingming Xu
- Department of Thoracic Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), 518020, Shenzhen, China
| | - Bin Peng
- Department of Thoracic Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), 518020, Shenzhen, China.
| |
Collapse
|
21
|
Chen Z, Yuan R, Hu S, Yuan W, Sun Z. Roles of the Exosomes Derived From Myeloid-Derived Suppressor Cells in Tumor Immunity and Cancer Progression. Front Immunol 2022; 13:817942. [PMID: 35154134 PMCID: PMC8829028 DOI: 10.3389/fimmu.2022.817942] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/12/2022] [Indexed: 12/23/2022] Open
Abstract
Tumor immunity is involved in malignant tumor progression. Myeloid-derived suppressor cells (MDSCs) play an irreplaceable role in tumor immunity. MDSCs are composed of immature myeloid cells and exhibit obvious immunomodulatory functions. Exosomes released by MDSCs (MDSCs-Exos) have similar effects to parental MDSCs in regulating tumor immunity. In this review, we provided a comprehensive description of the characteristics, functions and mechanisms of exosomes. We analyzed the immunosuppressive, angiogenesis and metastatic effects of MDSCs-Exos in different tumors through multiple perspectives. Immunotherapy targeting MDSCs-Exos has demonstrated great potential in cancers and non-cancerous diseases.
Collapse
Affiliation(s)
- Zhuang Chen
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rui Yuan
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shengyun Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weitang Yuan
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
22
|
Wang J, Li Y, Zhang C, Chen X, Zhu L, Luo T. A hypoxia-linked gene signature for prognosis prediction and evaluating the immune microenvironment in patients with hepatocellular carcinoma. Transl Cancer Res 2022; 10:3979-3992. [PMID: 35116696 PMCID: PMC8798548 DOI: 10.21037/tcr-21-741] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/30/2021] [Indexed: 12/22/2022]
Abstract
Background Previous research indicates that hypoxia critically affects the initiation and progression of hepatocellular carcinoma (HCC). Nevertheless, the molecular mechanisms responsible for HCC development are poorly understood. Herein, we purposed to build a prognostic model using hypoxia-linked genes to predict patient prognosis and investigate the relationship of hypoxia with immune status in the tumor microenvironment (TME). Methods The training cohort included transcriptome along with clinical data abstracted from The Cancer Genome Atlas (TCGA). The validation cohort was abstracted from Gene Expression Omnibus (GEO). Univariate along with multivariate Cox regression were adopted to create the prediction model. We divided all patients into low- and high-risk groups using median risk scores. The estimation power of the prediction model was determined with bioinformatic tools. Results Six hypoxia-linked genes, HMOX1, TKTL1, TPI1, ENO2, LDHA, and SLC2A1, were employed to create an estimation model. Kaplan-Meier, ROC curve, and risk plot analyses demonstrated that the estimation potential of the risk model was satisfactory. Univariate along with multivariate regression data illustrated that the risk model could independently predict the overall survival (OS). A nomogram integrating the risk signature and clinicopathological characteristics showed a good potential to estimate HCC prognosis. Gene set enrichment analysis (GSEA) revealed that genes associated with cell proliferation and metabolism cascades were abundant in high-risk group. Furthermore, the signature showed a strong ability to distinguish the two groups in terms of immune status. Conclusions A prediction model for predicting HCC prognosis using six hypoxia-linked genes was designed in this study, facilitating the diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Jukun Wang
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yu Li
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Chao Zhang
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xin Chen
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Linzhong Zhu
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Tao Luo
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, Beijing, China
| |
Collapse
|
23
|
Allam A, Yakou M, Pang L, Ernst M, Huynh J. Exploiting the STAT3 Nexus in Cancer-Associated Fibroblasts to Improve Cancer Therapy. Front Immunol 2021; 12:767939. [PMID: 34858425 PMCID: PMC8632218 DOI: 10.3389/fimmu.2021.767939] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/26/2021] [Indexed: 12/12/2022] Open
Abstract
The tumor microenvironment (TME) is composed of a heterogenous population of cells that exist alongside the extracellular matrix and soluble components. These components can shape an environment that is conducive to tumor growth and metastatic spread. It is well-established that stromal cancer-associated fibroblasts (CAFs) in the TME play a pivotal role in creating and maintaining a growth-permissive environment for tumor cells. A growing body of work has uncovered that tumor cells recruit and educate CAFs to remodel the TME, however, the mechanisms by which this occurs remain incompletely understood. Recent studies suggest that the signal transducer and activator of transcription 3 (STAT3) is a key transcription factor that regulates the function of CAFs, and their crosstalk with tumor and immune cells within the TME. CAF-intrinsic STAT3 activity within the TME correlates with tumor progression, immune suppression and eventually the establishment of metastases. In this review, we will focus on the roles of STAT3 in regulating CAF function and their crosstalk with other cells constituting the TME and discuss the utility of targeting STAT3 within the TME for therapeutic benefit.
Collapse
Affiliation(s)
- Amr Allam
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Marina Yakou
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Lokman Pang
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Jennifer Huynh
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| |
Collapse
|
24
|
Ma H, Wang J, Zhang XD. Near-infrared II emissive metal clusters: From atom physics to biomedicine. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214184] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
25
|
Liu J, Zheng Z, Zhang W, Wan M, Ma W, Wang R, Yan Y, Guo Y, Zhang J, Li W, Yao X. Dysregulation of tumor microenvironment promotes malignant progression and predicts risk of metastasis in bladder cancer. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1438. [PMID: 34733990 PMCID: PMC8506754 DOI: 10.21037/atm-21-4023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/02/2021] [Indexed: 11/12/2022]
Abstract
Background The tumor microenvironment (TME) is not only a key factor in the malignant progression of cancer but also plays an indispensable role in tumor immunotherapy. As an important regulatory factor in the TME, long non-coding RNAs (incRNA) are important for the development of bladder cancer. The purpose of this study was to explore the molecular mechanism of malignant progression of bladder cancer (BCa) from the perspective of immunology, establish a reliable signature, and evaluate its effect on prognosis, metastasis, and the effectiveness of immunotherapy. Methods The TME was assessed by single-sample gene set enrichment analysis (ssGSEA) in 373 patients with muscle invasive bladder cancer (MIBC) in The Cancer Genome Atlas (TCGA). Combining RNA sequence data from 49 BCa patients in our center, we established TME-related prognostic signatures (TMERPS) based on TME-related immune prognosis genes using weighted gene correlation network analysis, selection operator Cox analysis, minimum absolute shrinkage, and survival analysis. Real-Time Quantitative PCR was used for expression level analysis of related genes. Functional enrichment analysis and nomograms were used to explore the potential impact of TMERPS on the immune system, prognosis, and metastasis. Results The ssGSEA proved to be an accurate assessment of immune levels in BCa samples. TMERPS was established based on six TME-associated prognostic lncRNAs and was shown to be closely associated with prognosis, metastasis, and immune levels, and to have a significant stratifying effect on the therapeutic efficacy of immune checkpoint inhibitors. Finally, three TMERPS-based nomograms were shown to be effective in predicting prognosis, lymph node metastasis, and distant metastasis in BCa patients. Conclusions TMERPS can stratify BCa patients into different risk groups with different prognoses, immunotherapy sensitivity, and risk of metastasis. TMERPS-based nomograms can effectively predict prognosis and metastasis in BCa patients.
Collapse
Affiliation(s)
- Ji Liu
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Institute of Urinary Oncology, School of Medicine, Tongji University, Shanghai, China
| | - Zongtai Zheng
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Institute of Urinary Oncology, School of Medicine, Tongji University, Shanghai, China
| | - Wentao Zhang
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Institute of Urinary Oncology, School of Medicine, Tongji University, Shanghai, China
| | - Moxi Wan
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Institute of Urinary Oncology, School of Medicine, Tongji University, Shanghai, China
| | - Wenchao Ma
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Institute of Urinary Oncology, School of Medicine, Tongji University, Shanghai, China
| | - Ruiliang Wang
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Institute of Urinary Oncology, School of Medicine, Tongji University, Shanghai, China
| | - Yang Yan
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Institute of Urinary Oncology, School of Medicine, Tongji University, Shanghai, China
| | - Yadong Guo
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Institute of Urinary Oncology, School of Medicine, Tongji University, Shanghai, China
| | - Junfeng Zhang
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Institute of Urinary Oncology, School of Medicine, Tongji University, Shanghai, China
| | - Wei Li
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Institute of Urinary Oncology, School of Medicine, Tongji University, Shanghai, China
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Institute of Urinary Oncology, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
26
|
Aparicio C, Belver M, Enríquez L, Espeso F, Núñez L, Sánchez A, de la Fuente MÁ, González-Vallinas M. Cell Therapy for Colorectal Cancer: The Promise of Chimeric Antigen Receptor (CAR)-T Cells. Int J Mol Sci 2021; 22:11781. [PMID: 34769211 PMCID: PMC8583883 DOI: 10.3390/ijms222111781] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/23/2021] [Accepted: 10/26/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is a global public health problem as it is the third most prevalent and the second most lethal cancer worldwide. Major efforts are underway to understand its molecular pathways as well as to define the tumour-associated antigens (TAAs) and tumour-specific antigens (TSAs) or neoantigens, in order to develop an effective treatment. Cell therapies are currently gaining importance, and more specifically chimeric antigen receptor (CAR)-T cell therapy, in which genetically modified T cells are redirected against the tumour antigen of interest. This immunotherapy has emerged as one of the most promising advances in cancer treatment, having successfully demonstrated its efficacy in haematological malignancies. However, in solid tumours, such as colon cancer, it is proving difficult to achieve the same results due to the shortage of TSAs, on-target off-tumour effects, low CAR-T cell infiltration and the immunosuppressive microenvironment. To address these challenges in CRC, new approaches are proposed, including combined therapies, the regional administration of CAR-T cells and more complex CAR structures, among others. This review comprehensively summarises the current landscape of CAR-T cell therapy in CRC from the potential tumour targets to the preclinical studies and clinical trials, as well as the limitations and future perspectives of this novel antitumour strategy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Margarita González-Vallinas
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), University of Valladolid (UVa)-CSIC, 47003 Valladolid, Spain; (C.A.); (M.B.); (L.E.); (F.E.); (L.N.); (A.S.); (M.Á.d.l.F.)
| |
Collapse
|
27
|
Kiguchi T, Yamaguchi M, Takezako N, Miyawaki S, Masui K, Ihara Y, Hirota M, Shimofurutani N, Naoe T. Efficacy and safety of Wilms' tumor 1 helper peptide OCV-501 in elderly patients with acute myeloid leukemia: a multicenter, randomized, double-blind, placebo-controlled phase 2 trial. Cancer Immunol Immunother 2021; 71:1419-1430. [PMID: 34677647 DOI: 10.1007/s00262-021-03074-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 09/28/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE Complete remission (CR) of acute myeloid leukemia (AML) in elderly patients has a short duration, and there is no suitable post-remission therapy. We explored the role of the Wilms' tumor 1 helper peptide OCV-501 to prevent recurrence after remission. METHODS This placebo-controlled phase 2 study was designed to evaluate accurately the efficacy and immunogenicity of OCV-501 in elderly AML patients. Elderly AML patients who achieved first CR were randomly allocated to receive either OCV-501 (N = 69) or placebo (N = 65) once a week for eight weeks and then every two weeks until week 104. The primary endpoint was disease-free survival (DFS). RESULTS Nineteen (27.5%) patients in the OCV-501 group and 23 (35.4%) patients in the placebo group completed the study without relapse. The median DFS in the OCV-501 and placebo groups was 12.1 and 8.4 months, respectively (p = 0.7671, hazard ratio [95% confidence interval]: 0.933 [0.590, 1.477]). The major drug adverse reactions were injection-site reactions. Although treatment with OCV-501 did not prolong DFS for elderly AML patients, post hoc analysis found that immune responders to OCV-501 whose specific IgG was > 10,000 ng/mL (N = 16) and whose WT1-specific interferon-γ response was > 10 pg/mL (N = 26) had significantly longer overall survival compared with placebo. CONCLUSIONS The placebo-controlled design of this study and quantitative immunological monitoring provides new insight into the relationship between peptide-induced immune responses and survival, suggesting future perspectives for cancer immunotherapy.
Collapse
Affiliation(s)
| | | | - Naoki Takezako
- National Hospital Organization Disaster Medical Center of Japan, Tokyo, Japan
| | | | | | | | | | | | - Tomoki Naoe
- National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| |
Collapse
|
28
|
Wang DK, Zuo Q, He QY, Li B. Targeted Immunotherapies in Gastrointestinal Cancer: From Molecular Mechanisms to Implications. Front Immunol 2021; 12:705999. [PMID: 34447376 PMCID: PMC8383067 DOI: 10.3389/fimmu.2021.705999] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022] Open
Abstract
Gastrointestinal cancer is a leading cause of cancer-related mortality and remains a major challenge for cancer treatment. Despite the combined administration of modern surgical techniques and chemoradiotherapy (CRT), the overall 5-year survival rate of gastrointestinal cancer patients in advanced stage disease is less than 15%, due to rapid disease progression, metastasis, and CRT resistance. A better understanding of the mechanisms underlying cancer progression and optimized treatment strategies for gastrointestinal cancer are urgently needed. With increasing evidence highlighting the protective role of immune responses in cancer initiation and progression, immunotherapy has become a hot research topic in the integrative management of gastrointestinal cancer. Here, an overview of the molecular understanding of colorectal cancer, esophageal cancer and gastric cancer is provided. Subsequently, recently developed immunotherapy strategies, including immune checkpoint inhibitors, chimeric antigen receptor T cell therapies, tumor vaccines and therapies targeting other immune cells, have been described. Finally, the underlying mechanisms, fundamental research and clinical trials of each agent are discussed. Overall, this review summarizes recent advances and future directions for immunotherapy for patients with gastrointestinal malignancies.
Collapse
Affiliation(s)
| | | | | | - Bin Li
- Ministry of Education (MOE), Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, Jinan University, Guangzhou, China
| |
Collapse
|
29
|
Identification of prognostic immune-related gene signature associated with tumor microenvironment of colorectal cancer. BMC Cancer 2021; 21:905. [PMID: 34364366 PMCID: PMC8349485 DOI: 10.1186/s12885-021-08629-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 07/13/2021] [Indexed: 12/22/2022] Open
Abstract
Background The tumor microenvironment (TME) has significantly correlation with tumor occurrence and prognosis. Our study aimed to identify the prognostic immune-related genes (IRGs)in the tumor microenvironment of colorectal cancer (CRC). Methods Transcriptome and clinical data of CRC cases were downloaded from TCGA and GEO databases. Stromal score, immune score, and tumor purity were calculated by the ESTIMATE algorithm. Based on the scores, we divided CRC patients from the TCGA database into low and high groups, and the differentially expressed genes (DEGs) were identified. Immune-related genes (IRGs) were selected by venn plots. To explore underlying pathways, protein-protein interaction (PPI) networks and functional enrichment analysis were used. After utilizing LASSO Cox regression analysis, we finally established a multi-IRGs signature for predicting the prognosis of CRC patients. A nomogram consists of the thirteen-IRGs signature and clinical parameters was developed to predict the overall survival (OS). We investigated the association between prognostic validated IRGs and immune infiltrates by TIMER database. Results Gene expression profiles and clinical information of 1635 CRC patients were collected from the TCGA and GEO databases. Higher stromal score, immune score and lower tumor purity were observed positive correlation with tumor stage and poor OS. Based on stromal score, immune score and tumor purity, 1517 DEGs, 1296 DEGs, and 1892 DEGs were identified respectively. The 948 IRGs were screened by venn plots. A thirteen-IRGs signature was constructed for predicting survival of CRC patients. Nomogram with a C-index of 0.769 (95%CI, 0.717–0.821) was developed to predict survival of CRC patients by integrating clinical parameters and thirteen-IRGs signature. The AUC for 1-, 3-, and 5-year OS were 0.789, 0.783 and 0.790, respectively. Results from TIMER database revealed that CD1B, GPX3 and IDO1 were significantly related with immune infiltrates. Conclusions In this study, we established a novel thirteen immune-related genes signature that may serve as a validated prognostic predictor for CRC patients, thus will be conducive to individualized treatment decisions. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08629-3.
Collapse
|
30
|
Li X, Liu P, Sun X, Ma R, Cui T, Wang T, Bai Y, Li Y, Wu X, Feng X. Analyzing the impact of ATF3 in tumorigenesis and immune cell infiltration of ovarian tumor: a bioinformatics study. Med Oncol 2021; 38:91. [PMID: 34216322 DOI: 10.1007/s12032-021-01541-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/21/2021] [Indexed: 12/20/2022]
Abstract
ATF3 is an essential transcription activator in regulating cancer-related genetic expression. To identify the role of ATF3 in ovarian tumor, we investigated the correlation between ATF3 expression and the clinicopathological properties using multiple database. The cBioPortal and GEPIA database displayed the clinical information of ovarian patients harboring or without harboring ATF3 mutation. Furthermore, we assessed the relationship between survival and ATF3 expression level using Kaplan-Meier plotter, which reveals that the ovarian patients with higher expression of ATF3 suffered the worse overall survival and progression-free survival. The differentially expressed genes were analyzed using gene ontology, protein-protein interaction network, and gene set enrichment analysis to identify the hub gene and critical pathways, significantly affecting the tumorigenesis of ovarian tumor. Finally, we assessed the correlation between ATF3 and immune cell infiltration using Tumor Immunoassay Resource (TIMER) database. The results demonstrated that higher expression has a positive correlation with macrophage infiltration, expression for M1- and M2-type macrophages. Our study suggests that ATF3 can regulate the cell cycle and heme-related oxidative phosphorylation process, and it may be a critical factor to regulate the macrophage cell to be infiltrated into ovarian cancer. ATF3 can be used as a biomarker for diagnosis and therapy of ovarian tumor.
Collapse
Affiliation(s)
- Xiaoliu Li
- Department of Gynaecology, Henan Province People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Panpan Liu
- Department of Gynaecology, Henan Province People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Xiaona Sun
- Department of Gynaecology, Henan Province People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Runhong Ma
- Department of Gynaecology, Henan Province People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Ting Cui
- Department of Gynaecology, Henan Province People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Ting Wang
- Department of Gynaecology, Henan Province People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Yang Bai
- Department of Gynaecology, Henan Province People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Yuxia Li
- Department of Gynaecology, Henan Province People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Xiujuan Wu
- School of Life Sciences, Henan University, Kaifeng, 475000, Henan, China.
| | - Xianling Feng
- Department of Gynaecology, Henan Province People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China.
| |
Collapse
|
31
|
Galluzzi L, Garg AD. Immunology of Cell Death in Cancer Immunotherapy. Cells 2021; 10:cells10051208. [PMID: 34063358 PMCID: PMC8156735 DOI: 10.3390/cells10051208] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 05/13/2021] [Indexed: 12/22/2022] Open
Affiliation(s)
- Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, New York, NY 10065, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY 10065, USA
- Department of Dermatology, Yale School of Medicine, New Haven, CT 06520, USA
- Université de Paris, 75006 Paris, France
- Correspondence: (L.G.); (A.D.G.)
| | - Abhishek D. Garg
- Cell Stress & Immunity (CSI) Lab, Department for Cellular & Molecular Medicine (CMM), KU Leuven, 3000 Leuven, Belgium
- Correspondence: (L.G.); (A.D.G.)
| |
Collapse
|
32
|
Viralippurath Ashraf J, Sasidharan Nair V, Saleh R, Elkord E. Role of circular RNAs in colorectal tumor microenvironment. Biomed Pharmacother 2021; 137:111351. [PMID: 33550046 DOI: 10.1016/j.biopha.2021.111351] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 02/06/2023] Open
Abstract
Circular RNAs (circRNAs) are a class of endogenous noncoding RNA, which were previously considered as a byproduct of RNA splicing error. Numerous studies have demonstrated the altered expression of circRNAs in organ tissues during pathological conditions and their involvements in disease pathogenesis and progression, including cancers. In colorectal cancer (CRC), multiple circRNAs have been identified and characterized as "oncogenic", given their involvements in the downregulation of tumor suppressor genes and induction of tumor initiation, progression, invasion, and metastasis. Additionally, other circRNAs have been identified in CRC and characterized as "tumor suppressive" based on their ability of inhibiting the expression of oncogenic genes and suppressing tumor growth and proliferation. circRNAs could serve as potential diagnostic and prognostic biomarkers, and therapeutic targets or vectors to be utilized in cancer therapies. This review briefly describes the dynamic changes of the tumor microenvironment inducing immunosuppression and tumorigenesis, and outlines the biogenesis and characteristics of circRNAs and recent findings indicating their roles and functions in the CRC tumor microenvironment. It also discusses strategies and technologies, which could be employed in the future to overcome current cancer therapy challenges associated with circRNAs.
Collapse
Affiliation(s)
| | - Varun Sasidharan Nair
- Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Reem Saleh
- Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Eyad Elkord
- College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar; Biomedical Research Centre, School of Science, Engineering and Environment, University of Salford, Manchester, UK.
| |
Collapse
|
33
|
Lin Y, He Z, Ye J, Liu Z, She X, Gao X, Liang R. Progress in Understanding the IL-6/STAT3 Pathway in Colorectal Cancer. Onco Targets Ther 2020; 13:13023-13032. [PMID: 33376351 PMCID: PMC7762435 DOI: 10.2147/ott.s278013] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 12/01/2020] [Indexed: 12/19/2022] Open
Abstract
As a pleiotropic cytokine, interleukin-6 (IL-6) not only regulates the cellular immune response, but it also promotes tumor development by activating multiple carcinogenic pathways. IL-6 expression is significantly elevated in colorectal cancer (CRC) and is closely related to CRC development and patient prognosis. In CRC, IL-6 activates signal transducers and activators of transduction-3 (STAT3) to promote tumor initiation and tumor growth. IL-6/STAT3 signalling has a profound effect on tumor-infiltrating immune cells in the tumor immune microenvironment in CRC. Additionally, IL-6/STAT3 pathway activates downstream target genes to protect tumor cells from apoptosis; drive tumor cell proliferation, cell cycle progression, invasion and metastasis; promote tumor angiogenesis; and stimulate drug resistance. Therefore, a thorough understanding of the many effects of the IL-6/STAT3 pathway in CRC is needed, which the present review examines.
Collapse
Affiliation(s)
- Yan Lin
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, People's Republic of China
| | - Ziqin He
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, People's Republic of China
| | - Jiazhou Ye
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, People's Republic of China
| | - Ziyu Liu
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, People's Republic of China
| | - Xiaomin She
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, People's Republic of China
| | - Xing Gao
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, People's Republic of China
| | - Rong Liang
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, People's Republic of China
| |
Collapse
|
34
|
Yin K, Xia X, Rui K, Wang T, Wang S. Myeloid-Derived Suppressor Cells: A New and Pivotal Player in Colorectal Cancer Progression. Front Oncol 2020; 10:610104. [PMID: 33384962 PMCID: PMC7770157 DOI: 10.3389/fonc.2020.610104] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) remains a devastating human malignancy with poor prognosis. Of the various factors, immune evasion mechanisms play pivotal roles in CRC progression and impede the effects of cancer therapy. Myeloid-derived suppressor cells (MDSCs) constitute an immature population of myeloid cells that are typical during tumor progression. These cells have the ability to induce strong immunosuppressive effects within the tumor microenvironment (TME) and promote CRC development. Indeed, MDSCs have been shown to accumulate in both tumor-bearing mice and CRC patients, and may therefore become an obstacle for cancer immunotherapy. Consequently, numerous studies have focused on the characterization of MDSCs and their immunosuppressive capacity, as well as developing novel approaches to suppress MDSCs function with different approaches. Current therapeutic strategies that target MDSCs in CRC include inhibition of their recruitment and alteration of their function, alone or in combination with other therapies including chemotherapy, radiotherapy and immunotherapy. Herein, we summarize the recent roles and mechanisms of MDSCs in CRC progression. In addition, a brief review of MDSC-targeting approaches for potential CRC therapy is presented.
Collapse
Affiliation(s)
- Kai Yin
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xueli Xia
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Ke Rui
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Tingting Wang
- Department of Laboratory Medicine, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Children's Hospital, Wuxi, China
| | - Shengjun Wang
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
35
|
Guo Q, Li X, Cui MN, Sun JL, Ji HY, Ni BB, Yan MX. CD13: A Key Player in Multidrug Resistance in Cancer Chemotherapy. Oncol Res 2020; 28:533-540. [PMID: 32532363 PMCID: PMC7751223 DOI: 10.3727/096504020x15919605976853] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cancer is one of the most serious diseases that are harmful to human health. Systemic chemotherapy is an optimal therapeutic strategy for the treatment of cancer, but great difficulty has been encountered in its administration in the form of multidrug resistance (MDR). As an enzyme on the outer cell surface, CD13 is documented to be involved in the MDR development of tumor cells. In this review, we will focus on the role of CD13 in MDR generation based on the current evidence.
Collapse
Affiliation(s)
- Qie Guo
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao UniversityQingdao, ShandongP.R. China
| | - Xiao Li
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao UniversityQingdao, ShandongP.R. China
| | - Meng-Na Cui
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao UniversityQingdao, ShandongP.R. China
| | - Jia-Lin Sun
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao UniversityQingdao, ShandongP.R. China
| | - Hong-Yan Ji
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao UniversityQingdao, ShandongP.R. China
| | - Bei-Bei Ni
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao UniversityQingdao, ShandongP.R. China
| | - Mei-Xing Yan
- Department of Pharmacy, Qingdao Women and Childrens HospitalQingdao, ShandongP.R. China
| |
Collapse
|
36
|
Feng M, Zhao Z, Yang M, Ji J, Zhu D. T-cell-based immunotherapy in colorectal cancer. Cancer Lett 2020; 498:201-209. [PMID: 33129958 DOI: 10.1016/j.canlet.2020.10.040] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/08/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) is the leading cause of cancer death worldwide. CRC therapeutic strategies include surgical resection, chemotherapy, radiotherapy, and other approaches. However, patients with metastatic CRC have worse prognoses. In recent years, T-cell-based immunotherapy has elicited promising responses in B-cell malignancies, melanoma, and lung cancer, but most CRC patients are resistant to immunotherapy, chemotherapy, and targeted therapy. Immune checkpoint inhibitors have shown encouraging results in non-small cell lung cancer, melanoma, and other cancers, but immune checkpoint blockade is only effective for CRC subset with microsatellite instability. Other immunotherapies, such as cytokines, cancer vaccines, small molecules, oncolytic viruses, and chimeric antigen-receptor therapy, are currently in use against CRC. This review analyzes recent developments in immunotherapy for CRC treatment as well as the challenges in overcoming resistance.
Collapse
Affiliation(s)
- Mei Feng
- Minhang Hospital and Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Zhongwei Zhao
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Affiliated Lishui Hospital of Zhejiang University, Lishui, 323000, China
| | - Mengxuan Yang
- Minhang Hospital and Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Affiliated Lishui Hospital of Zhejiang University, Lishui, 323000, China.
| | - Di Zhu
- Minhang Hospital and Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China; Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai, 201203, China; Shanghai Engineering Research Center of ImmunoTherapeutics, Fudan University, 201203, China.
| |
Collapse
|
37
|
Chiang JH, Tsai FJ, Hsu YM, Yin MC, Chiu HY, Yang JS. Sensitivity of allyl isothiocyanate to induce apoptosis via ER stress and the mitochondrial pathway upon ROS production in colorectal adenocarcinoma cells. Oncol Rep 2020; 44:1415-1424. [PMID: 32700751 PMCID: PMC7448487 DOI: 10.3892/or.2020.7700] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023] Open
Abstract
Allyl isothiocyanate (AITC), a bioactive phytochemical compound that is a constituent of dietary cruciferous vegetables, possesses promising chemopreventive and anticancer effects. However, reports of AITC exerting antitumor effects on apoptosis induction of colorectal cancer (CRC) cells in vitro are not well elucidated. The present study focused on the functional mechanism of the endoplasmic reticulum (ER) stress-based apoptotic machinery induced by AITC in human colorectal cancer HT-29 cells. Our results indicated that AITC decreased cell growth and number, reduced viability, and facilitated morphological changes of apoptotic cell death. DNA analysis by flow cytometry showed G2/M phase arrest, and alterations in the modulated protein levels caused by AITC were detected via western blot analysis. AITC also triggered vital intrinsic apoptotic factors (caspase-9/caspase-3 activity), disrupted mitochondrial membrane potential, and stimulated mitochondrial-related apoptotic molecules (e.g., cytochrome c, apoptotic protease activating factor 1, apoptosis-inducing factor, and endonuclease G). Additionally, AITC prompted induced cytosolic Ca2+ release and Ca2+-dependent ER stress-related signals, such as calpain 1, activating transcription factor 6α, glucose-regulated proteins 78 and 94, growth arrest- and DNA damage-inducible protein 153 (GADD153), and caspase-4. The level of reactive oxygen species (ROS) production was found to induce the hallmark of ER stress GADD153, proapoptotic marker caspase-3, and calpain activity after AITC treatment. Our findings showed for the first time that AITC induced G2/M phase arrest and apoptotic death via ROS-based ER stress and the intrinsic pathway (mitochondrial-dependent) in HT-29 cells. Overall, AITC may exert an epigenetic effect and is a potential bioactive compound for CRC treatment.
Collapse
Affiliation(s)
- Jo-Hua Chiang
- Department of Nursing, Chung‑Jen Junior College of Nursing, Health Sciences and Management, Chiayi County 62241, Taiwan, R.O.C
| | - Fuu-Jen Tsai
- Human Genetics Center, Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan, R.O.C
| | - Yuan-Man Hsu
- Department of Biological Science and Technology, China Medical University, Taichung 40402, Taiwan, R.O.C
| | - Mei-Chin Yin
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung 41354, Taiwan, R.O.C
| | - Hong-Yi Chiu
- Department of Pharmacy, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97002, Taiwan, R.O.C
| | - Jai-Sing Yang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40442, Taiwan, R.O.C
| |
Collapse
|
38
|
Wang X, Wu Y, Wen D, Wu LY, Zhao Y, He Y, Yang H. An Individualized Immune Prognostic Index is a Superior Predictor of Survival of Hepatocellular Carcinoma. Med Sci Monit 2020; 26:e921786. [PMID: 32527991 PMCID: PMC7285951 DOI: 10.12659/msm.921786] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background The tumor microenvironment is largely orchestrated by the immune cells. Considerable evidence has shown their excellent clinicopathological application value in assessment of clinical outcomes and immunotherapy efficacy. Hence, a moderate, individualized prognostic signature based on immune cells that can estimate prognosis and reflect the immune microenvironment in hepatocellular carcinoma (HCC) patients is greatly needed. Material/Methods Here, we systematically analyzed the expression differences and survival prediction value of tumor infiltrating immune cells by analyzing 638 HCC patients from 3 public cohorts, including 2 microarray datasets and 1 RNA sequencing dataset. CIBERSORT software, a computational algorithm, was used to calculate the relative levels of immune cells. Three immune microenvironment subtypes were defined via ConsensuClusterPlus package. Univariate and multivariate survival analyses were used to develop an individualized immune prognostic index based on immune cell pairs. Results Notably, HCC patients with higher immune signatures score, utterly appreciable, suffered inferior prognosis (hazard ratio=2.742; 95% confidence interval: 1.887–3.983; P<0.001). Subgroup analysis suggested that the prognostic signature did particularly well in early-stage patients. Furthermore, moderate survival prediction value was also confirmed in another two independent cohorts GSE14520 and GSE76427. Conclusions This study provides a systematic view of the immune cells characteristics in HCC and suggests their superior survival monitoring performance.
Collapse
Affiliation(s)
- Xiaodong Wang
- Department of Medical Ultrasonics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Yuquan Wu
- Department of Medical Ultrasonics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Dongyue Wen
- Department of Medical Ultrasonics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Lin-Yong Wu
- Department of Medical Ultrasonics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Yujia Zhao
- Department of Medical Ultrasonics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Yun He
- Department of Medical Ultrasonics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Hong Yang
- Department of Medical Ultrasonics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| |
Collapse
|
39
|
|
40
|
An Y, Yang Q. MiR-21 modulates the polarization of macrophages and increases the effects of M2 macrophages on promoting the chemoresistance of ovarian cancer. Life Sci 2019; 242:117162. [PMID: 31837336 DOI: 10.1016/j.lfs.2019.117162] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 11/06/2019] [Accepted: 12/09/2019] [Indexed: 01/07/2023]
Abstract
AIMS Chemoresistance is a major underlying cause of relapse or death in ovarian cancer patients. Emerging evidence has shown that macrophages could play an essential role in mediating the chemoresistance of cancer cells. MiR-21 has been reported to be an oncogene, which promotes chemoresistance in cancer. Here, we aim to investigate the role that miR-21 plays in polarization of macrophages and ovarian cancer progression. MAIN METHODS The CIBERSORT algorithm was used to investigate immune cell infiltration in ovarian cancer tissues. To explore the role that miR-21 played in macrophages, M2 macrophages transfected with a miR-21 mimic or a miR-21 inhibitor were co-cultured with ovarian cancer cells. Western blotting was used to detect protein expression levels. CCK8 was used to detect the IC50 of ovarian cancer cells. Flow cytometry was used to detect apoptosis and the cell cycle of ovarian cancer cells. KEY FINDINGS In this study, we found that higher expression of M1 macrophages and lower expression of M2 macrophages correlated with a better prognosis of ovarian cancer patients. M2 macrophages promoted the chemoresistance of ovarian cancer cells. The results showed that miR-21 could partially regulate the polarization of macrophages. Furthermore, M2 macrophages transfected with the miR-21 mimic significantly promoted chemoresistance and inhibited apoptosis of ovarian cancer cells, while the M2 macrophages transfected with the miR-21 inhibitor showed the opposite effects. SIGNIFICANCE miR-21 plays an important role in regulating macrophage polarization, therefore increasing the M2 macrophage-mediated chemoresistance in ovarian cancer cells.
Collapse
Affiliation(s)
- Yuanyuan An
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, PR China
| | - Qing Yang
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, PR China.
| |
Collapse
|